WO1998052951A1 - Novel triptolide derivatives useful in the treatment of autoimmune diseases - Google Patents

Novel triptolide derivatives useful in the treatment of autoimmune diseases Download PDF

Info

Publication number
WO1998052951A1
WO1998052951A1 PCT/US1998/008470 US9808470W WO9852951A1 WO 1998052951 A1 WO1998052951 A1 WO 1998052951A1 US 9808470 W US9808470 W US 9808470W WO 9852951 A1 WO9852951 A1 WO 9852951A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compound according
amino acid
formula
suitable amino
Prior art date
Application number
PCT/US1998/008470
Other languages
English (en)
French (fr)
Inventor
Michael J. Jung
Mahinda Wickramaratne
Michael Hepperle
Original Assignee
Hoechst Marion Roussel, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to EP98919928A priority Critical patent/EP0983275B1/de
Priority to DK98919928T priority patent/DK0983275T3/da
Application filed by Hoechst Marion Roussel, Inc. filed Critical Hoechst Marion Roussel, Inc.
Priority to NZ500762A priority patent/NZ500762A/en
Priority to CA002287977A priority patent/CA2287977A1/en
Priority to DE69807466T priority patent/DE69807466T2/de
Priority to KR19997010786A priority patent/KR20010012822A/ko
Priority to IL13308898A priority patent/IL133088A0/xx
Priority to AU72607/98A priority patent/AU741917B2/en
Priority to AT98919928T priority patent/ATE222910T1/de
Priority to JP55037798A priority patent/JP2001525851A/ja
Priority to HU0003398A priority patent/HUP0003398A3/hu
Priority to BR9809678-8A priority patent/BR9809678A/pt
Priority to SI9830161T priority patent/SI0983275T1/xx
Publication of WO1998052951A1 publication Critical patent/WO1998052951A1/en
Priority to NO995723A priority patent/NO995723L/no
Priority to HK00105108A priority patent/HK1026096A1/xx

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/12Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains three hetero rings
    • C07D493/14Ortho-condensed systems

Definitions

  • autoimmune diseases such as rheumatoid arthritis, nephritis, uveitis, thyroiditis, and early stage of insulin dependent diabetes mellitus, systemic lupus erythematosus, psoriasis and inflammatory bowel disease.
  • the immune system when operating normally is involved in precise functions such as recognition and memory of, specific response to, and clearance of, foreign substances (chemical and cellular antigens) that either penetrate the protective body barriers of skin and mucosal surfaces (transplanted tissue and microorganisms such as bacteria, viruses, parasites) or arise de novo (malignant transformation).
  • the arsenal of the immune response is composed of two major types of lymphocytes that are either B-lymphocytes (B cells, responsible for producing antibodies which attack the invading microorganisms) or the T-lymphocytes (T cells, responsible for eliminating the infected or abnormal target cells) in cooperation with macrophages.
  • B cells B-lymphocytes
  • T cells responsible for eliminating the infected or abnormal target cells
  • the response is initiated by the interaction of an antigen with macrophages and surface antibodies on B cells.
  • the macrophages ingest and process the antigen.
  • the activated macrophages secrete interleukin-1 (IL-1) and tumor necrosis factor (TNF), and display the processed antigen on the cell surface together with a major antihistocompatibility antigen.
  • IL-1 and TNF initiate a number of processes involving inflammation.
  • IL-1 induces proliferation of B cells and synthesis of antibodies.
  • IL-1 activates T cells which release a series of lymphokines including interleukin-2 (IL-2) that activate the proliferation of T cells and cytotoxic lymphocytes.
  • IL-2 interleukin-2
  • Each element in the cascade of the immune response may be considered as a potential site for pharmacological intervention.
  • adrenocorticosteroids act in the first stages of the immune response, interact with the macrophages and, inhibit the synthesis and release of IL-1.
  • Other immunosuppressive agents used in the treatment of autoimmune diseases have been identified, such as azathioprine and methotrexate for rheumatoid arthritis, cyclophosphamide for nephritic conditions of immune origin, and cyclosporin for rheumatoid arthritis, uveitis, early onset insulin dependent diabetes mellitus, psoriasis, nephritic syndrome and aplastic anemia.
  • immunosuppressive agents have proved to be useful in preventing and treating organ transplantation rejection that may occur in allograft transplantation.
  • allograft transplantation one person donates an organ to a genetically disparate individual while in xenograft transplantation an organ of one species is transplanted into a member of another species.
  • the use of cyclosporin has shown a real improvement in the condition of the person receiving the organ.
  • the therapeutic index of the available immunosuppressive drugs is narrow, none of the drugs are completely effective and their use has been limited by severe toxicity.
  • T wilfordii Hook F an herbal plant from the Celastraceae family that is grown mainly in the southern part of China.
  • Zhang, et al., Shanghai Yike Da ue Xuebao, 13(4), 267 (1986) have characterized T wilfordii as comprising at least six different diterpenoids, including triptonide, triptolide, triptophenolide and triptonolide. More specifically, P.E. Lipsky, et al.
  • Triptolide has been reported by Yang, et al, Int. J. Immunopharmac, 14, 963 (1992) and Yang, et al., Int. J. Immunopharmac, 16, 895 (1994), to suppress lymphocyte proliferation and skin allograft rejection.
  • Jin and Wiedmann in WO 94/26265, published November 24, 1994, disclosed a composition wherein an additional component of Tripterygium wilfordii Hook F, purified 16-hydroxytriptolide, is administered in conjunction with another immunosuppressive agent, such as cyclosporin A, FK506, azathioprine, methotrexate, rapamycin, mycophenolic acid, or a glucocorticoid.
  • Another immunosuppressive agent such as cyclosporin A, FK506, azathioprine, methotrexate, rapamycin, mycophenolic acid, or a glucocorticoid.
  • the above composition was disclosed as providing an increase in immunosuppressive activity relative to the sum of the effects produced by 16- hydroxytriptolide or the other immunosuppressive agent used alone.
  • Ri and R 2 are each independently is H or -OR 5 ;
  • R 4 is H or -OH
  • the present invention further relates to novel compounds of the formula (II):
  • X is I, Br, Cl, F or -CN
  • Ri and R 2 are each independently is H or -OR 5 ;
  • R 4 is H or -OH;
  • the present invention relates to novel compounds of the formula
  • Ri and R 2 are each independently is H or -OR 5 ;
  • R 4 is H or -OH
  • the present invention further provides novel intermediates for the preparation of compounds of formulas (I), (II) and (III).
  • the present invention provides a method of treating a patient suffering from an autoimmune disease comprising administering to a patient an effective amount of a compound of formulas (I), (II) or (III).
  • stereoisomers is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes geometric (cis/trans) isomers, and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers).
  • chiral center refers to a carbon atom to which four different groups are attached.
  • enantiomer or “enantiomeric” refers to a molecule that is nonsuperimposable on its mirror image and hence optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image rotates the plane of polarized light in the opposite direction.
  • racemic mixture or “racemic modification” refers to a mixture of equal parts of enantiomers and which is optically inactive.
  • prefixes "(+)” and “(-)” are employed to designate the sign of rotation of the plane of polarized light by the compound, with (+) meaning the compound is dextrorotatory and (-) meaning the compound is levorotatory.
  • LJD amino-acids
  • R/S can be used as described in IUPAC-IUB Joint Commission on Biochemical Nomenclature, Eur. J. Biochem. 138: 9-37 (1984).
  • “Pharmaceutically acceptable salt” is a non-toxic organic or inorganic acid addition salt of the base compounds represented by formulas (I), (II) or (III), or any of their intermediates.
  • inorganic acids which form suitable salts include hydrochloric, hydrobromic, sulphuric, and phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate, and potassium hydrogen sulfate.
  • Illustrative organic acids which form suitable salts include the mono-, di-, and tricarboxylic acids.
  • Such acids are acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, hydroxymaleic, benzoic, hydroxybenzoic, phenylacetic, cinnamic, salicyclic, 2- phenoxybenzoic, p-toluenesulfonic acid, and sulfonic acids such as methane sulfonic acid and 2-hyroxyethane sulfonic acid.
  • Such salts can exist in either a hydrated or substantially anhydrous form.
  • enantiomeric enrichment refers to the increase in the amount of one enantiomer as compared to its corresponding opposite enantiomer.
  • a convenient method of expressing enantiomeric enrichment achieved is the concept of "enantiomeric excess” or "ee”, which is expressed by the following equation;
  • E 1 is the amount of the first enantiomer and E 2 is the amount of the second corresponding enantiomer.
  • E 2 is the amount of the second corresponding enantiomer.
  • the designation " - — * " refers to a bond that protrudes forward out of the plane of the page.
  • «w refers to a bond that protrudes backward out of the plane of the page.
  • wvw refers to a single or double bond.
  • triptolide in its optically active form and racemic form is readily available to one of ordinary skill in the art.
  • Triptolide, in optically active form can be isolated from the natural source Tripterygium wilfordii following the procedure of S. M. Kupchan, et al., J. Am. Chem. Soc, 94, 7194 (1972).
  • triptolide can be prepared in its racemic form following the total synthesis of Chee Kong Lai, et al., J. Org. Chem., 47, 2364-2369 (1982), van Tamelen and Leiden, J. Am. Chem.
  • Triptolidenol and “15-Hydroxytriptolide” are the same have the following structure;
  • Tripterinin and “16-Hydroxytriptolide” are the same and have the following structure;
  • each a-amino acid has a characteristic "R-group", the R-group being the side chain, or residue, attached to the a-carbon atom of the a-amino acid.
  • R-group side chain for glycine is hydrogen, for alanine it is methyl, for valine it is isopropyl.
  • R-groups or side chains of the a-amino acids see A.L. Lehninger's text on Biochemistry.
  • the a-amino acids utilized in the compounds of the present invention are preferably in their L-configu ration; however, applicants contemplate that the amino acids employed herein can be of either the D- or L- configurations or can be mixtures of the D- and L-isomers, including the racemic mixture.
  • the recognized abbreviations for the a-amino acids are set forth in Table I.
  • suitable amino acid refers to the amino acids listed in Table I above.
  • the suitable amino acid is connected to the compound of formulas (I), (II) or (III) at the carboxy end of the amino acid, resulting in an ester linkage.
  • glutamic acid at the -OH at position 14 of formula (I) provides a compound with the following structure:
  • Preferred suitable amino acids are Ala, Gin, Lys, Arg, and Phe, with Ala, Gin and Lys being most preferred.
  • step A the compound of structure (1) is subjected to cis-diol formation conditions to produce the compound of formula (la) wherein R 1a and R 2a are each independently hydrogen or -OH.
  • the compound of structure (1) such as triptolide is dissolved in a suitable anhydrous organic solvent, such as tetrahydrofuran under an inert atmosphere, such as nitrogen at room temperature.
  • a suitable anhydrous organic solvent such as tetrahydrofuran under an inert atmosphere, such as nitrogen at room temperature.
  • the solution is treated with about 1 to about 6 equivalents of sodium cyanoborohydride followed by dropwise addition of about 1.2 equivalents of neat boron trifluoride diethyl etherate (BF 3 Et 2 O).
  • the reaction mixture is allowed to stir for about 1 hours to about 24 hours, with about 16 hours being preferred.
  • the reaction is then quenched by addition of aqueous ammonium chloride and the product is isolated and purified by techniques well known in the art, such as extraction
  • the quenched reaction mixture is extracted with a suitable organic solvent, such as methylene chloride.
  • a suitable organic solvent such as methylene chloride.
  • the organic extracts are combined, rinsed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the crude product.
  • the crude product is then purified by flash chromatography on silica gel with a suitable eluent, such as methanol/chloroform to provide the purified compound of formula (la).
  • the compound of formula (la) can be prepared by dissolving the compound of structure (1 ) in a suitable anhydrous organic solvent, such as tetrahydrofuran under an inert atmosphere, such as nitrogen, at room temperature. To this is added about 1.9 equivalents of lithium borohydride followed by addition of about 2.1 equivalents of neat boron trifluoride diethyl etherate. The reaction mixture is allowed to stir for about 0.5 hours to about 5 hours, with about 1.5 hours being preferred. The reaction is then carefully quenched with 1 N HCI and the product is isolated and purified by techniques well known in the art, such as extraction techniques and chromatography.
  • a suitable anhydrous organic solvent such as tetrahydrofuran under an inert atmosphere, such as nitrogen
  • the quenched reaction is extracted with a suitable organic solvent, such as methylene chloride.
  • a suitable organic solvent such as methylene chloride.
  • the organic extracts are combined, rinsed with 1 N sodium bicarbonate, brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the crude product.
  • the crude product is then purified by flash chromatography on silica gel with a suitable eluent, such as ethyl acetate/hexane to provide the purified compound of formula (la).
  • step B the compound of formula (la) is subjected to standard acylation conditions well known to one of ordinary skill in the art to provide the compound of formula (lb).
  • a suitable organic solvent such as dimethylformamide.
  • the solution is then treated with one equivalent of a suitable cyclic anhydride, such as succinic anhydride, one equivalent of 4-dimethylaminopyridine (DMAP) and about 1.6 equivalents of pyridine.
  • DMAP 4-dimethylaminopyridine
  • the compound of formula (lb) is isolated and purified by techniques well known in the art such as extraction techniques and chromatography. For example, the residue is dissolved in a suitable organic solvent, such as methylene chloride, rinsed with water, brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the crude product. The crude product is then purified by flash chromatography on silica gel with a suitable eluent, such as methanol/chloroform to provide the purified compound of formula (lb).
  • a suitable organic solvent such as methylene chloride
  • a suitable eluent such as methanol/chloroform
  • the compound of formula (la) can be dissolved in a suitable anhydrous organic solvent, such as pyridine, and then treated with the acid chloride of a suitable monoester of the corresponding suitable diacid.
  • suitable diacids are succinic acid, glutaric acid, adipic acid, pimelic acid and suberic acid.
  • a suitable monoester of the diacid is an ester that is ultimately de-esterified under mild conditions well known in the art to provide the compound of formula (lb).
  • the above solution is treated with one equivalent of the acid chloride of mono-tert-butyl succinate or mono-benzyl succinate, and then stirred for about 2 hours at room temperature.
  • the resulting ester is then isolated and purified by techniques well known in the art. For example, the solvent is removed under vacuum and the residue is purified by flash chromatography on silica gel with a suitable eluent, such as methanol/chloroform to provide the purified ester.
  • the ester is then converted to the corresponding acid by techniques well known in the art such as acid hydrolysis of the tert-butyl ester or hydrogenation of the benzyl ester as disclosed by T.W. Green, "Protective Groups in Organic Synthesis", John Wiley & Sons Inc, 1981 , pages 168-169 and 171-172 respectively to provide the crude compound of formula (lb).
  • the crude material is then purified and mixtures separated if necessary by flash chromatography on silica gel with a suitable eluent, such as methanol/chloroform to provide the purified compound of formula (lb).
  • the compound of formula (la) must be treated with at least 1.2 equivalents of the suitable cyclic anhydride, at least 1.2 equivalents of DMAP and at least 1.8 equivalents of pyridine, for every primary and secondary alcohol present on the compound of formula (la).
  • the compound of formula (la) can be acylated with an amino acid from Table I, which has been suitably protected, under conditions well known in the art, followed by deprotection of the amino acid portion of the compound to provide the compound of formula (lb).
  • Table I which has been suitably protected, under conditions well known in the art, followed by deprotection of the amino acid portion of the compound to provide the compound of formula (lb).
  • each amino acid to be coupled to the primary or secondary alcohol must be protected.
  • Any protecting group known in the art can be used. Examples of which include: 1) acyl types such as formyl, trifluoroacetyl, phthalyl, and p-toluenesulfonyl; 2) aromatic carbamate types such as benzyloxycarbonyl (Cbz or Z) and substituted benzyloxycarbonyls, 1-(p-biphenyl)-1- methylethoxy-carbonyl, and 9-fluorenylmethyloxycarbonyl (Fmoc); 3) aliphatic carbamate types such as tert-butyloxycarbonyl (Boc), ethoxycarbonyl, diisopropyl- methoxycarbonyl, and allyloxycarbonyl; 4) cyclic alkyl carbamate types such as cyclopentyloxycarbonyi and adamantyloxycarbonyl;
  • the a-amino protecting group of the added amino acid residue is then cleaved under conditions well known in the art to provide the compound of formula (lb).
  • Boc group the methods of choice are trifluoroacetic acid, neat or in methylene chloride, or HCI in dioxane or ethyl acetate as disclosed in Greene "Protective Groups in Organic Synthesis", John Wiley & Sons, New York (1981), 232-233.
  • the compound of formula (la) is dissolved in a suitable anhydrous organic solvent, such as methylene choride under an inert atmosphere, such as nitrogen, and treated with an excess of a suitably protected amino acid and about two equivalents of dimethylaminopyridine.
  • suitably protected amino acids are N-(tert-butoxycarbonyl)-L-alanine, N-(tert-butoxycarbonyl)-L- phenylalanine, N,N'-(ditert-butoxycarbonyl)-lysine, N-(t-butylcarbonyl)omega tertbutoxyl-glutamate and the like.
  • the solution is cooled to 0°C with stirring and treated with about two equivalents of dicyclohexylcarbodiimide. Then allow the reaction to warm to room temperature and stir for about 3 hours. Then filter the reaction to remove any precipitate and concentrate the filtrate under vacuum.
  • the BOC protected compound of formula (lb) dissolve the BOC protected compound of formula (lb) in a suitable organic solvent, such as diethyl ether and treat the solution slowly with 1 N trifluoroacetic acid. Allow the reaction to stir for about 1 hour.
  • the trifluoro acetate salt of the compound of formula (lb) is then collected by filtration, washed with diethyl ether and dried.
  • the free base of the compound of formula (lb) can be prepared by dissolving the trifluoroacetate salt of formula (lb) in water and treating with at least one equivalent of sodium carbonate.
  • the aqueous solution is then extracted with a suitable organic solvent, such as methylene chloride.
  • the organic extract is then washed with water, brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the compound of formula (lb).
  • step C the 12-13 epoxide of formula (la) is opened to provide the compound of formula (lla).
  • the compound of formula (la) is dissolved in a suitable organic solvent, such as dioxane at room temperature and an excess of a suitable aqueous acid, such as 2N HCI or 30% aqueous HBr is added.
  • a suitable aqueous acid such as 2N HCI or 30% aqueous HBr is added.
  • the reaction is allowed to stir at a temperature of about 23°C to about 70°C for about 1 hour to about 24 hours, with about 6 hours being preferred.
  • the product is then isolated and purified by techniques well known in the art, such as extraction techniques and chromatography.
  • the reaction is diluted with water and extracted with a suitable organic solvent, such as ethyl acetate, methylene chloride or chloroform.
  • a suitable organic solvent such as ethyl acetate, methylene chloride or chloroform.
  • the organic extracts are combined, washed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the crude product of formula (lla).
  • the crude product is then purified by flash chromatography on silica gel with a suitable eluent, such as methanol/chloroform or hexane/methylene chloride to provide the purified compound of formula (lla).
  • step D the compound of formula (lla) is subjected to standard acylation conditions in a manner analogous to the procedures described in Scheme A, step B, to provide the compound of formula (lib).
  • step A the compound of structure (1) is subjected to epoxide opening conditions.
  • the compound of structure (1 ) is dissolved in a suitable organic solvent, such as acetone or dioxane and treated with a suitable aqueous acid, such as 2N HCI or 30% HBr.
  • a suitable organic solvent such as acetone or dioxane
  • the reaction is then diluted with water and the compound of structure (2) is isolated and purified by techniques well known in the art, such as extraction techniques, chromatography and/or recrystallization.
  • the reaction mixture is extracted with a suitable organic solvent, such as methylene chloride or ethyl acetate.
  • the organic extracts are combined, rinsed with brine, dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide the crude product (2).
  • the crude product is then purified by recrystallization from a suitable solvent system, such as hexane/methylene chloride to provide the purified compound of structure (2).
  • step B the compound of structure (2) is converted to the ether of formula (Ilia) wherein R ⁇ a and R 2a are each independently hydrogen or -OH.
  • compound (2) is combined with finely powdered potassium hydrogen sulfate in a suitable anhydrous organic solvent, such as toluene or dioxane.
  • a suitable anhydrous organic solvent such as toluene or dioxane.
  • potassium hydrogen sulfate can be replaced by 0.1 to 1 N aqueous HCI and the preferred organic solvent is dioxane.
  • the reaction mixture is then heated at about 50°C to about 115°C, with about 75°C being preferred for about 1 hour to about 24 hours, with about 5 hours being preferred.
  • the reaction is then filtered, for example through diatomaceous earth, and the solids are rinsed with a suitable organic solvent, such as chloroform.
  • a suitable organic solvent such as chloroform.
  • the filtrate is then concentrated under vacuum to provide the crude compound of formula (Ilia).
  • the crude material is then purified by techniques well known in the art, such as chromatography on silica gel with a suitable eluent, such as methanol/chloroform to provide the purified compound of formula (Ilia).
  • compound (2) can be converted to the compound of formula (Ilia) by treatment of compound (2) with a suitable Lewis acid, such as BFa/etherate, TiCI 4 or FeCI 3 in a suitable organic solvent, such as methylene chloride for about 1 to 2 hours at room temperature.
  • a suitable Lewis acid such as BFa/etherate, TiCI 4 or FeCI 3
  • a suitable organic solvent such as methylene chloride
  • step C the compound of formula (Ilia) is acylated in a manner analogous to the procedure previously described in Scheme A, step B to provide the compound of formula (1Mb).
  • the reaction mixture is stirred for 5 hours and then is diluted with cold water (5 mL).
  • the mixture is extracted with ethyl acetate (3 X 10 mL), the organic extracts are combined, washed with brine (3 X 10 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum.
  • the residue is purified by column chromatography (silica gel, 40% ethyl acetate/hexane) to provide the title compound (4.8 mg, 82%).
  • step A Triptolide (100 mg, 0.277 mmol, optically active, isolated from natural source) is combined with a mixture of dioxane (15 mL) and 1.5N aqueous HCI (15 mL), and the reaction mixture is stirred for 48 hours at room temperature. The reaction mixture is then poured into water (50 mL) and extracted with chloroform (3 X 40 mL). The combined organic extracts are washed with brine (60 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum to provide a white solid (116 mg). This white solid is then recrystallized from hexane/chloroform to provide the title compound (65 mg) as white needles.
  • reaction mixture is then poured into water (5 mL) and extracted with chloroform ( 3 X 5 mL). The organic extracts are combined, washed with brine (5 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The residue is purified by preparative TLC (20 X 20 cm, 0.25 mm, 5% methanol/chloroform) to provide the title compound (4 mg, 36%).
  • step A Tripdiolide (0.277 mmol, optically active, isolated from natural source) is combined with a mixture of dioxane (15 mL) and 1.5N aqueous HCI (15 mL), and the reaction mixture is stirred for 48 hours at room temperature. The reaction mixture is then poured into water (50 mL) and extracted with chloroform (3 X 40 mL). The combined organic extracts are washed with brine (60 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The residue is then purified by flash chromatography (silica gel, 2% methanol/chloroform) to provide the title compound.
  • reaction mixture is then poured into water (5 mL) and extracted with chloroform ( 3 X 5 mL). The organic extracts are combined, washed with brine (5 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The residue is purified by preparative TLC (20 X 20 cm, 0.25 mm, 3% methanol/chloroform) to provide the title compound.
  • step A Triptolidenol (100 mg) is combined with a mixture of dioxane (15 mL) and 1.5N aqueous HCI (15 mL), and the reaction mixture is stirred for 48 hours at room temperature. The reaction mixture is then poured into water (50 mL) and extracted with chloroform (3 X 40 mL). The combined organic extracts are washed with brine (60 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The residue is purified by flash chromatography (silica gel, 2% methanol/chloroform) to provide the title compound.
  • reaction mixture is then poured into water (5 mL) and extracted with chloroform (3 X 5 mL). The organic extracts are combined, washed with brine (5 mL), dried over anhydrous magnesium sulfate, filtered and concentrated under vacuum. The residue is purified by preparative TLC (20 X 20 cm, 0.25 mm, 3% methanol/chloroform) to provide the title compound.
  • Jurkat E6-1 cell line a human leukemic T cell line or lymphocytes prepared from fresh human blood, are grown in complete culture medium consisting of RPMI-1640 (Mediatech) supplemented with 10% v/v fetal bovine serum (HyClone, Logen, Vermont), penicillin (100 units/mL), streptomycin (200 mg/mL), and 2mM L- glutamine (GIBCO, Grand Island, New York).
  • the cells are grown to a concentration of between 1.2 to 1.8 X 10 6 cells/mL before use.
  • the cells are then centrifuged at low speed and resuspended to a concentration of 1.25 X 10 6 cells/mL in fresh medium.
  • the compounds to be tested are dissolved in DMSO, and water is added to make a 50% DMSO/water solution.
  • the compounds are then diluted in sterile water such that the DMSO concentrations are less then 0.05%.
  • the cells at a concentration of 1.25 X 10 6 cells/mL are stimulated with phytohemagglutinin 10 ⁇ g/mL (PHA) and 10 "8 M phorbol ester (12-O-tetradecanoylphorbol 13-acetate: TPA), (Sigma, St. Louis, Missouri).
  • PHA phytohemagglutinin 10 ⁇ g/mL
  • 10 "8 M phorbol ester (12-O-tetradecanoylphorbol 13-acetate: TPA) (Sigma, St. Louis, Missouri).
  • the compounds are then added and the cells are plated in flat-bottom tissue culture plates (Falcon, Lincoln Park, New Jersey), with 250,000 cells/well. Plates are then incubated at 37°C in a 5% CO
  • IL-2 production is assayed utilizing a solid phase ELISA immunoassay, sold in kit form by R&D Systems, Inc., Minneapolis, Minnesota.
  • MTT cell viability
  • Cell viability is measured by the use of a mitochondrial enzyme assay. After removal of the supernatant for the IL-2 assay, Cell Titer 96 (a commercial solution of MTT obtained from Promega, Madison, Wisconsin), is added to the plates and incubated for 4 hours at 37°C. A solubilization solution is then added and incubated overnight. The plates are then read on an ELISA plate reader at a wavelength of 570 nm. Table II presents the results for the lnterleukin-2 assay, the cell viability assay and the ratio of cell viability/I L-2 inhibition.
  • the present invention provides a method of treating a patient suffering from an autoimmune disease comprising administering to said patient an effective amount of a compound of either formula (I), (II) or (III).
  • autoimmune disease refers to those disease states and conditions wherein the immune response of the patient is directed against the patient's own constituents resulting in an undesirable and often knowingly debilitating condition. Included within the scope of autoimmune disease is ARDS, inflammatory bowel disease including ulcerative colitis and Crohn's disease, rheumatoid arthritis, diabetes mellitus type I, Kawasaki disease, multiple sclerosis, familial Mediterranean fever, psoriasis and lupus.
  • Patients suffering from autoimmune diseases are in need of treatment with an antiinflammatory agent such as a compound of formulas (I), (II) or (III).
  • patients suffering from allograft rejection and Graft-versus-host disease are also in need of treatment with an antiinflammatory agent such as a compound of formulas (I), (II) or (III).
  • an antiinflammatory agent such as a compound of formulas (I), (II) or (III).
  • treatment of patients suffering from these diseases by administration of a compound of formulas (I), (II) or (III) will be particularly effective in preventing further deterioration or worsening of the patient's condition.
  • Treatment of a patient at an early stage of an autoimmune disease would be particularly effective in preventing further deterioration of the disease state into a more serious condition.
  • autoimmune diseases for which treatment with a compound of formulas (I), (II) or (III) will be particularly preferred are rheumatoid arthritis, juvenile arthritis, systemic lupus erythematosus and psoriasis.
  • the term "patient” refers to a warm-blooded animal such as a mammal which is suffering from, or is in danger of suffering from, an acute or chronic inflammation, cellular injury or cell death associated with an immunological based disease, such as an autoimmune disease. It is understood that humans, dogs, guinea pigs, mice and rats are included within the scope of the term "patient”.
  • An effective amount of a compound of formulas (I), (II) or (III) is that amount which is effective, upon single or multiple dose administration to a patient, in providing an immunosuppressive or antiinflammatory effect.
  • an effective amount of a compound of formulas (I), (II) or (III) can be readily determined by the attending diagnostician, as one skilled in the art, by the use of known techniques and by observing results obtained under analogous circumstances. In determining the effective amount or dose, a number of factors are considered by the attending diagnostician, including, but not limited to: the species of mammal; its size, age, and general health; the specific disease involved; the degree of or involvement or the severity of the disease; the response of the individual patient; the particular compound administered; the mode of administration; the bioavailability characteristics of the preparation administered; the dose regimen selected; the use of concomitant medication; and other relevant circumstances.
  • An effective amount of a compound of formulas (I), (II) or (III) is expected to vary from about 0.1 milligram per kilogram of body weight per day (mg/kg/day) to about 50 mg/kg/day. Preferred amounts are expected to vary from about 0.2 to about 25 mg/kg/day, with about 1 to about 10 mg/kg/day being most preferred.
  • a compound of formulas (I), (II) or (III) can be administered in any form or mode which makes the compound bioavailable in effective amounts, including oral and parenteral routes.
  • compounds of formulas (I), (II) or (III) can be administered orally, subcutaneously, intramuscularly, intravenously, transdermally, intranasally, rectally, and the like. Oral administration and intravenous administration are generally preferred.
  • One skilled in the art of preparing formulations can readily select the proper form and mode of administration depending upon the particular characteristics of the compound selected the disease state to be treated, the stage of the disease, and other relevant circumstances.
  • the compounds of formulas (I), (II) or (III) can be administered alone or in the form of a pharmaceutical composition in combination with pharmaceutically acceptable carriers or excipients, the proportion and nature of which are determined by the solubility and chemical properties of the compound selected, the chosen route of administration, and standard pharmaceutical practice.
  • the compounds of the invention while effective themselves, may be formulated and administered in the form of their pharmaceutically acceptable acid addition salts for purposes of stability, convenience of crystallization, increased solubility and the like.
  • compositions comprising a compound of formulas (I), (II) or (III) in admixture or otherwise in association with one or more inert carriers.
  • These compositions are useful, for example, as assay standards, as convenient means of making bulk shipments, or as pharmaceutical compositions.
  • An assayable amount of a compound of formulas (I), (II) or (III) is an amount which is readily measurable by standard assay procedures and techniques as are well known and appreciated by those skilled in the art.
  • Assayable amounts of a compound of formulas (I), (II) or (III) will generally vary from about 0.001% to about 75% of the composition by weight.
  • Inert carriers can be any material which does not degrade or otherwise covalently react with a compound of formulas (I), (II) or (III).
  • suitable inert carriers are water; aqueous buffers, such as those which are generally useful in High Performance Liquid Chromatography (HPLC) analysis; organic solvents, such as acetonitrile, ethyl acetate, hexane and the like; and pharmaceutically acceptable carriers or excipients.
  • the present invention provides pharmaceutical compositions comprising an effective amount of a compound of formula (I), (II) or (III) in admixture or otherwise in association with one or more pharmaceutically acceptable carriers or excipients.
  • the pharmaceutical compositions are prepared in a manner well known in the pharmaceutical art.
  • the carrier or excipient may be a solid, semi-solid, or liquid material which can serve as a vehicle or medium for the active ingredient. Suitable carriers or excipients are well known in the art.
  • the pharmaceutical composition may be adapted for oral or parenteral use, including topical use, and may be administered to the patient in the form of tablets, capsules, suppositories, solution, suspensions, or the like.
  • the compounds of the present invention may be administered orally, for example, with an inert diluent or with an edible carrier. They may be enclosed in gelatin capsules or compressed into tablets.
  • the compounds may be incorporated with excipients and used in the form of tablets, troches, capsules, elixirs, suspensions, syrups, wafers, chewing gums and the like.
  • These preparations should contain at least 4% of the compound of the invention, the active ingredient, but may be varied depending upon the particular form and may conveniently be between 4% to about 70% of the weight of the unit.
  • the amount of the compound present in compositions is such that a suitable dosage will be obtained.
  • Preferred compositions and preparations according to the present invention are prepared so that an oral dosage unit form contains between 5.0-300 milligrams of a compound of the invention.
  • the tablets, pills, capsules, troches and the like may also contain one or more of the following adjuvants: binders such as microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch or lactose, disintegrating agents such as alginic acid, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; and sweetening agents such as sucrose or saccharin may be added or a flavoring agent such as peppermint, methyl salicylate or orange flavoring.
  • a liquid carrier such as polyethylene glycol or a fatty oil.
  • dosage unit forms may contain other various materials which modify the physical form of the dosage unit, for example, as coatings.
  • tablets or pills may be coated with sugar, shellac, or other enteric coating agents.
  • a syrup may contain, in addition to the present compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors. Materials used in preparing these various compositions should be pharmaceutically pure and non-toxic in the amounts used.
  • the compounds of the present invention may be incorporated into a solution or suspension. These preparations should contain at least 0.1% of a compound of the invention, but may be varied to be between 0.1 and about 50% of the weight thereof.
  • compositions and preparations according to the present invention are prepared so that a parenteral dosage unit contains between 5.0 to 100 milligrams of the compound of the invention.
  • the solutions or suspensions may also include the one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylene diaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the parenteral preparation can be enclosed in ampules, disposable syringes or multiple dose vials made of glass or plastic.
PCT/US1998/008470 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases WO1998052951A1 (en)

Priority Applications (15)

Application Number Priority Date Filing Date Title
AT98919928T ATE222910T1 (de) 1997-05-23 1998-04-27 Triptolid-derivate nützlich zur behandlung von autoimmunkrankheiten
AU72607/98A AU741917B2 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases
NZ500762A NZ500762A (en) 1997-05-23 1998-04-27 Triptolide derivatives substituted with amino acids, useful in the treatment of autoimmune diseases
DK98919928T DK0983275T3 (da) 1997-05-23 1998-04-27 Nye triptolidderivater, der er nyttige til behandling af autoimmune sygdomme
DE69807466T DE69807466T2 (de) 1997-05-23 1998-04-27 Triptolid-derivate nützlich zur behandlung von autoimmunkrankheiten
KR19997010786A KR20010012822A (ko) 1997-05-23 1998-04-27 자가 면역 질환의 치료에 유용한 신규의 트리프톨라이드유도체
JP55037798A JP2001525851A (ja) 1997-05-23 1998-04-27 自己免疫疾患の治療に有用な新規なトリプトリド誘導体
EP98919928A EP0983275B1 (de) 1997-05-23 1998-04-27 Triptolid-derivate nützlich zur behandlung von autoimmunkrankheiten
CA002287977A CA2287977A1 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases
IL13308898A IL133088A0 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases
HU0003398A HUP0003398A3 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives and pharmaceutical compositions thereof
BR9809678-8A BR9809678A (pt) 1997-05-23 1998-04-27 Derivados de triptolìdeo úteis no tratamento de doenças autoimunes
SI9830161T SI0983275T1 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases
NO995723A NO995723L (no) 1997-05-23 1999-11-22 Nye triptolidderivater som er anvendbare ved behandling av autoimmune sykdommer
HK00105108A HK1026096A1 (en) 1997-05-23 2000-08-16 Novel triptolide derivatives useful in the treatment of autoimmune diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US86248897A 1997-05-23 1997-05-23
US08/862,488 1997-05-23

Publications (1)

Publication Number Publication Date
WO1998052951A1 true WO1998052951A1 (en) 1998-11-26

Family

ID=25338616

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/008470 WO1998052951A1 (en) 1997-05-23 1998-04-27 Novel triptolide derivatives useful in the treatment of autoimmune diseases

Country Status (21)

Country Link
EP (1) EP0983275B1 (de)
JP (1) JP2001525851A (de)
KR (1) KR20010012822A (de)
CN (1) CN1257507A (de)
AR (1) AR012723A1 (de)
AT (1) ATE222910T1 (de)
AU (1) AU741917B2 (de)
BR (1) BR9809678A (de)
CA (1) CA2287977A1 (de)
DE (1) DE69807466T2 (de)
DK (1) DK0983275T3 (de)
ES (1) ES2178207T3 (de)
HK (1) HK1026096A1 (de)
HU (1) HUP0003398A3 (de)
IL (1) IL133088A0 (de)
NO (1) NO995723L (de)
NZ (1) NZ500762A (de)
PT (1) PT983275E (de)
TW (1) TW434235B (de)
WO (1) WO1998052951A1 (de)
ZA (1) ZA984174B (de)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6329148B1 (en) 1999-02-16 2001-12-11 The Board Of Trustees Of The Leland Stanford University Combined therapy of diterpenoid triepoxides and death domain receptor ligands for synergistic killing of tumor cells
EP1390358A1 (de) * 2001-03-15 2004-02-25 Pharmagenesis, Inc. Aminosäurederivate von triptolidverbindungen als immunmodulatoren und antikrebsmittel
EP1511478A2 (de) * 2002-05-31 2005-03-09 Pharmagenesis, Inc. Triptolid-derivate zur modulierung der apoptose und immunosuppression
US7820834B2 (en) 2003-12-24 2010-10-26 Pharmagenesis, Inc. Triptolide 5,6-derivatives as immunomodulators and anticancer agents
US7863464B2 (en) 2004-03-02 2011-01-04 Pharmagenesis, Inc. Triptolide lactone ring derivatives as immunomodulators and anticancer agents
US8048914B2 (en) 2004-02-09 2011-11-01 Pharmagenesis, Inc. Methods for isolation of triptolide compounds from Tripterygium wilfordii
JP2012516899A (ja) * 2009-02-05 2012-07-26 ファーマジェネシス, インコーポレイテッド 抗癌剤および免疫調節因子としてのトリプトリドc環誘導体
EP2583678A2 (de) 2004-06-24 2013-04-24 Novartis Vaccines and Diagnostics, Inc. Immunstimulatoren kleiner Moleküle und Assays für deren Nachweis
US8617906B2 (en) 2004-10-13 2013-12-31 Pharmagenesis, Inc. Identification and screening of triptolide target molecules
EP3248981A4 (de) * 2015-10-29 2018-07-18 Hangzhou Weben Pharmaceuticals Inc. C14-hydroxylveresterte aminosäurederivate von triptolid sowie herstellungsverfahren und verwendung davon
CN114716503A (zh) * 2022-03-23 2022-07-08 广东省中医院(广州中医药大学第二附属医院、广州中医药大学第二临床医学院、广东省中医药科学院) 一种雷公藤氯内酯醇的制备方法

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100398544C (zh) * 2002-09-18 2008-07-02 成都达远药物有限公司 高免疫抑制活性的水溶性雷公藤内酯醇衍生物及其应用
CN1726025A (zh) * 2002-12-17 2006-01-25 泛华医药公司 作为免疫调节剂和抗癌剂的雷公藤内酯醇衍生物
CN1223595C (zh) 2002-12-27 2005-10-19 中国科学院上海药物研究所 雷公藤内酯醇衍生物及其应用
US6943259B2 (en) * 2003-02-25 2005-09-13 Pharmagenesis, Inc. Halogenated triptolide derivatives as immunomodulators and anticancer agents

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4005108A (en) * 1973-04-03 1977-01-25 Research Corporation Novel anti-leukemic diterpenoid triepoxides
US5430054A (en) * 1989-12-22 1995-07-04 Jiangsu Family Planning Institute Preparation methods of diterpene lactone compounds and application of the same to antifertility
US5663335A (en) * 1996-03-01 1997-09-02 Pharmagenesis, Inc. Immunosuppressive compounds and methods

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4005108A (en) * 1973-04-03 1977-01-25 Research Corporation Novel anti-leukemic diterpenoid triepoxides
US5430054A (en) * 1989-12-22 1995-07-04 Jiangsu Family Planning Institute Preparation methods of diterpene lactone compounds and application of the same to antifertility
US5663335A (en) * 1996-03-01 1997-09-02 Pharmagenesis, Inc. Immunosuppressive compounds and methods

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, vol. 124, no. 19, 6 May 1996, Columbus, Ohio, US; abstract no. 255764y, PENG-CHEN MA ET AL: "Neotriptetraolide isolated from Tripterygium wilfordii Hook. F." page 733; XP002073335 *
ZHIWU XUEBAO, vol. 37, no. 10, 1995, pages 822 - 828 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6329148B1 (en) 1999-02-16 2001-12-11 The Board Of Trustees Of The Leland Stanford University Combined therapy of diterpenoid triepoxides and death domain receptor ligands for synergistic killing of tumor cells
EP1390358A1 (de) * 2001-03-15 2004-02-25 Pharmagenesis, Inc. Aminosäurederivate von triptolidverbindungen als immunmodulatoren und antikrebsmittel
EP1390358A4 (de) * 2001-03-15 2004-06-16 Pharmagenesis Inc Aminosäurederivate von triptolidverbindungen als immunmodulatoren und antikrebsmittel
EP2468277A1 (de) * 2002-05-31 2012-06-27 Pharmagenesis, Inc. Triptolidderivate zur Modulierung der Apoptose und Immunsuppression
EP1511478A2 (de) * 2002-05-31 2005-03-09 Pharmagenesis, Inc. Triptolid-derivate zur modulierung der apoptose und immunosuppression
US7662976B2 (en) 2002-05-31 2010-02-16 Pharmagenesis, Inc. Triptolide derivatives for modulation of apoptosis and immunosuppression
US7847109B2 (en) 2002-05-31 2010-12-07 Pharmagenesis, Inc. Triptolide derivatives for modulation of apoptosis and immunosuppression
EP1511478A4 (de) * 2002-05-31 2009-12-02 Pharmagenesis Inc Triptolid-derivate zur modulierung der apoptose und immunosuppression
US7820834B2 (en) 2003-12-24 2010-10-26 Pharmagenesis, Inc. Triptolide 5,6-derivatives as immunomodulators and anticancer agents
US8048914B2 (en) 2004-02-09 2011-11-01 Pharmagenesis, Inc. Methods for isolation of triptolide compounds from Tripterygium wilfordii
US8426616B2 (en) 2004-03-02 2013-04-23 Pharmagenesis, Inc. Triptolide lactone ring derivatives as immunomodulators and anticancer agents
US7863464B2 (en) 2004-03-02 2011-01-04 Pharmagenesis, Inc. Triptolide lactone ring derivatives as immunomodulators and anticancer agents
EP2583678A2 (de) 2004-06-24 2013-04-24 Novartis Vaccines and Diagnostics, Inc. Immunstimulatoren kleiner Moleküle und Assays für deren Nachweis
US8617906B2 (en) 2004-10-13 2013-12-31 Pharmagenesis, Inc. Identification and screening of triptolide target molecules
JP2012516899A (ja) * 2009-02-05 2012-07-26 ファーマジェネシス, インコーポレイテッド 抗癌剤および免疫調節因子としてのトリプトリドc環誘導体
EP3248981A4 (de) * 2015-10-29 2018-07-18 Hangzhou Weben Pharmaceuticals Inc. C14-hydroxylveresterte aminosäurederivate von triptolid sowie herstellungsverfahren und verwendung davon
US10238623B2 (en) 2015-10-29 2019-03-26 Hangzhou Weben Pharmaceuticals Inc C14-hydroxyl esterified amino acid derivative of triptolide, and preparation method and use thereof
AU2015413013B2 (en) * 2015-10-29 2020-11-05 Hangzhou Weben Pharmaceuticals Inc C14-hydroxyl esterified amino acid derivatives of triptolide, and preparation method and use thereof
CN114716503A (zh) * 2022-03-23 2022-07-08 广东省中医院(广州中医药大学第二附属医院、广州中医药大学第二临床医学院、广东省中医药科学院) 一种雷公藤氯内酯醇的制备方法
CN114716503B (zh) * 2022-03-23 2024-03-12 广东省中医院(广州中医药大学第二附属医院、广州中医药大学第二临床医学院、广东省中医药科学院) 一种雷公藤氯内酯醇的制备方法

Also Published As

Publication number Publication date
PT983275E (pt) 2003-01-31
NO995723L (no) 2000-01-21
ATE222910T1 (de) 2002-09-15
NZ500762A (en) 2001-08-31
EP0983275B1 (de) 2002-08-28
DK0983275T3 (da) 2002-12-23
ES2178207T3 (es) 2002-12-16
KR20010012822A (ko) 2001-02-26
HUP0003398A2 (en) 2001-03-28
CA2287977A1 (en) 1998-11-26
DE69807466T2 (de) 2003-04-24
CN1257507A (zh) 2000-06-21
HK1026096A1 (en) 2000-12-01
BR9809678A (pt) 2000-07-11
EP0983275A1 (de) 2000-03-08
AU741917B2 (en) 2001-12-13
ZA984174B (en) 1998-11-23
IL133088A0 (en) 2001-03-19
NO995723D0 (no) 1999-11-22
HUP0003398A3 (en) 2001-09-28
AU7260798A (en) 1998-12-11
DE69807466D1 (de) 2002-10-02
JP2001525851A (ja) 2001-12-11
TW434235B (en) 2001-05-16
AR012723A1 (es) 2000-11-08

Similar Documents

Publication Publication Date Title
US6004999A (en) Triptolide derivatives useful in the treatment of autoimmune diseases
EP0983275B1 (de) Triptolid-derivate nützlich zur behandlung von autoimmunkrankheiten
JPH0556357B2 (de)
US5972998A (en) Triptolide derivatives useful in the treatment of autoimmune diseases
AU764123B2 (en) Triptolide prodrugs having high aqueous solubility
EP0983256B1 (de) Triptolid-derivate geeignet zur behandlung von autoimmunkrankheiten
JPH08503940A (ja) 酵素阻害薬
US6569893B2 (en) Amino acid derivatives of triptolide compounds as immune modulators and anticancer agents
CA2163348C (en) Calanolide and related antiviral compounds, compositions, and uses thereof
JP2002537284A (ja) メビノリン誘導体
WO2021027306A1 (zh) 双氢青蒿素甾体缀合物及其制备方法和应用
CN1185222C (zh) 用于治疗自身免疫性疾病的新型雷公藤内酯衍生物
CN108003027B (zh) 1-o-咖啡酰奎宁酸、其衍生物、制备方法及其用途
FR2483929A1 (fr) Nouvelles adenosines n6-substituees utiles comme medicaments antihypertenseurs, compositions therapeutiques et formes pharmaceutiques les contenant, et leur procede de preparation
JP2716572B2 (ja) アシルアミノカルボン酸誘導体、その製法及びこれを含有する薬剤及び食餌療法剤
WO2021047524A1 (zh) 一类靶向蛋白质水解通路的功能分子及其制备和应用
EP0467280B1 (de) Spergulinähnliche Verbindungen und ihre Verwendung
CN117105895A (zh) 火把花烷二倍半萜colquhounoid D衍生物及其制备方法和应用
KR20150034068A (ko) 백선피 추출물과 이로부터 분리된 신규 글래브레탈-타입 트리테르페노이드 및 이의 용도
JPH05178806A (ja) オキシアミノ酸残基を含む新規ペプタイドおよびそれを含有する免疫増強剤
JPH0466582A (ja) マクロライド物質
JP2004161728A (ja) ナキテルピオシン誘導体及びこれを有効成分とする抗癌剤
WO1997001334A1 (en) Cytotoxic triterpenes from a marine sponge

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 133088

Country of ref document: IL

Ref document number: 98805397.7

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM GW HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2287977

Country of ref document: CA

Ref document number: 2287977

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 500762

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 72607/98

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 1998 550377

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: PA/a/1999/010776

Country of ref document: MX

Ref document number: 1019997010786

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 1998919928

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1998919928

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1019997010786

Country of ref document: KR

WWG Wipo information: grant in national office

Ref document number: 72607/98

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: 1998919928

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1019997010786

Country of ref document: KR